Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
RNA ; 30(8): 955-966, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38777382

ABSTRACT

The long noncoding RNA TERRA is transcribed from telomeres in virtually all eukaryotes with linear chromosomes. In humans, TERRA transcription is driven in part by promoters comprising CpG dinucleotide-rich repeats of 29 bp repeats, believed to be present in half of the subtelomeres. Thus far, TERRA expression has been analyzed mainly using molecular biology-based approaches that only generate partial and somehow biased results. Here, we present a novel experimental pipeline to study human TERRA based on long-read sequencing (TERRA ONTseq). By applying TERRA ONTseq to different cell lines, we show that the vast majority of human telomeres produce TERRA and that the cellular levels of TERRA transcripts vary according to their chromosomes of origin. Using TERRA ONTseq, we also identified regions containing TERRA transcription start sites (TSSs) in more than half of human subtelomeres. TERRA TSS regions are generally found immediately downstream from 29 bp repeat-related sequences, which appear to be more widespread than previously estimated. Finally, we isolated a novel TERRA promoter from the highly expressed subtelomere of the long arm of Chromosome 7. With the development of TERRA ONTseq, we provide a refined picture of human TERRA biogenesis and expression and we equip the scientific community with an invaluable tool for future studies.


Subject(s)
Promoter Regions, Genetic , RNA, Long Noncoding , Telomere , Transcription Initiation Site , Transcriptome , Humans , Telomere/genetics , Telomere/metabolism , RNA, Long Noncoding/genetics , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods
2.
FEBS Lett ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38445359

ABSTRACT

Eukaryotic telomeres are transcribed into the long noncoding RNA TERRA. A fraction of TERRA remains associated with telomeres by forming RNA:DNA hybrids dubbed telR-loops. TERRA and telR-loops are essential to promote telomere elongation in human cancer cells that maintain telomeres through a homology-directed repair pathway known as alternative lengthening of telomeres or ALT. However, TERRA and telR-loops compromise telomere integrity and cell viability if their levels are not finely tuned. The study of telomere transcription in ALT cells will enormously expand our understanding of the ALT mechanism and of how genome integrity is maintained. Moreover, telomere transcription, TERRA and telR-loops are likely to become exceptionally suited targets for the development of novel anti-cancer therapies.

4.
Int J Mol Sci ; 23(18)2022 Sep 09.
Article in English | MEDLINE | ID: mdl-36142374

ABSTRACT

RNA-protein interactions drive key cellular pathways such as protein translation, nuclear organization and genome stability maintenance. The human telomeric protein TRF2 binds to the long noncoding RNA TERRA through independent domains, including its N-terminal B domain. We previously demonstrated that TRF2 B domain binding to TERRA supports invasion of TERRA into telomeric double stranded DNA, leading to the formation of telomeric RNA:DNA hybrids. The other telomeric protein TRF1, which also binds to TERRA, suppresses this TRF2-associated activity by preventing TERRA-B domain interactions. Herein, we show that the binding of both TRF1 and TRF2 to TERRA depends on the ability of the latter to form G-quadruplex structures. Moreover, a cluster of arginines within the B domain is largely responsible for its binding to TERRA. On the other side, a patch of glutamates within the N-terminal A domain of TRF1 mainly accounts for the inhibition of TERRA-B domain complex formation. Finally, mouse TRF2 B domain binds to TERRA, similarly to its human counterpart, while mouse TRF1 A domain lacks the inhibitory activity. Our data shed further light on the complex crosstalk between telomeric proteins and RNAs and suggest a lack of functional conservation in mouse.


Subject(s)
RNA, Long Noncoding , Telomeric Repeat Binding Protein 1 , Telomeric Repeat Binding Protein 2 , Animals , DNA/chemistry , Glutamates , Humans , Mice , RNA, Long Noncoding/genetics , Telomere/genetics , Telomere/metabolism , Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 1/genetics , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/genetics
5.
Proc Natl Acad Sci U S A ; 119(39): e2208669119, 2022 09 27.
Article in English | MEDLINE | ID: mdl-36122232

ABSTRACT

A substantial number of human cancers are telomerase-negative and elongate physiologically damaged telomeres through a break-induced replication (BIR)-based mechanism known as alternative lengthening of telomeres (ALT). We recently demonstrated that inhibiting the transcription of the telomeric long noncoding RNA TERRA suppresses telomere damage and ALT features, indicating that telomere transcription is a main trigger of ALT activity. Here we show that experimentally increased TERRA transcription not only increases ALT features, as expected, but also causes rapid loss of telomeric DNA through a pathway that requires the endonuclease Mus81. Our data indicate that the ALT mechanism can endanger telomere integrity if not properly controlled and point to TERRA transcription as a uniquely versatile target for therapy.


Subject(s)
RNA, Long Noncoding , Telomerase , DNA , Endonucleases/metabolism , Humans , Telomerase/genetics , Telomere/genetics , Telomere/metabolism
6.
Elife ; 112022 02 22.
Article in English | MEDLINE | ID: mdl-35191837

ABSTRACT

DNA oxidation by ten-eleven translocation (TET) family enzymes is essential for epigenetic reprogramming. The conversion of 5-methylcytosine (5mC) into 5-hydroxymethylcytosine (5hmC) initiates developmental and cell-type-specific transcriptional programs through mechanisms that include changes in the chromatin structure. Here, we show that the presence of 5hmC in the transcribed gene promotes the annealing of the nascent RNA to the template DNA strand, leading to the formation of an R-loop. Depletion of TET enzymes reduced global R-loops in the absence of gene expression changes, whereas CRISPR-mediated tethering of TET to an active gene promoted the formation of R-loops. The genome-wide distribution of 5hmC and R-loops shows a positive correlation in mouse and human stem cells and overlap in half of all active genes. Moreover, R-loop resolution leads to differential expression of a subset of genes that are involved in crucial events during stem cell proliferation. Altogether, our data reveal that epigenetic reprogramming via TET activity promotes co-transcriptional R-loop formation, disclosing new mechanisms of gene expression regulation.


Subject(s)
Dioxygenases , R-Loop Structures , 5-Methylcytosine/metabolism , Animals , Cytosine , DNA/metabolism , DNA Methylation , Dioxygenases/genetics , Epigenesis, Genetic , Epigenomics , Humans , Mice
7.
Nucleic Acids Res ; 50(1): 350-367, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34928380

ABSTRACT

Although the RNA helicase Upf1 has hitherto been examined mostly in relation to its cytoplasmic role in nonsense mediated mRNA decay (NMD), here we report high-throughput ChIP data indicating genome-wide association of Upf1 with active genes in Schizosaccharomyces pombe. This association is RNase sensitive, correlates with Pol II transcription and mRNA expression levels. Changes in Pol II occupancy were detected in a Upf1 deficient (upf1Δ) strain, prevalently at genes showing a high Upf1 relative to Pol II association in wild-type. Additionally, an increased Ser2 Pol II signal was detected at all highly transcribed genes examined by ChIP-qPCR. Furthermore, upf1Δ cells are hypersensitive to the transcription elongation inhibitor 6-azauracil. A significant proportion of the genes associated with Upf1 in wild-type conditions are also mis-regulated in upf1Δ. These data envisage that by operating on the nascent transcript, Upf1 might influence Pol II phosphorylation and transcription.


Subject(s)
RNA Helicases/metabolism , RNA Polymerase II/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Gene Expression Regulation, Fungal , Genome, Fungal , Phosphorylation , RNA Helicases/genetics , RNA Polymerase II/genetics , Schizosaccharomyces , Schizosaccharomyces pombe Proteins/genetics , Transcriptional Activation
8.
Nat Commun ; 12(1): 3760, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145295

ABSTRACT

Alternative Lengthening of Telomeres (ALT) is a Break-Induced Replication (BIR)-based mechanism elongating telomeres in a subset of human cancer cells. While the notion that spontaneous DNA damage at telomeres is required to initiate ALT, the molecular triggers of this physiological telomere instability are largely unknown. We previously proposed that the telomeric long noncoding RNA TERRA may represent one such trigger; however, given the lack of tools to suppress TERRA transcription in cells, our hypothesis remained speculative. We have developed Transcription Activator-Like Effectors able to rapidly inhibit TERRA transcription from multiple chromosome ends in an ALT cell line. TERRA transcription inhibition decreases marks of DNA replication stress and DNA damage at telomeres and impairs ALT activity and telomere length maintenance. We conclude that TERRA transcription actively destabilizes telomere integrity in ALT cells, thereby triggering BIR and promoting telomere elongation. Our data point to TERRA transcription manipulation as a potentially useful target for therapy.


Subject(s)
RNA, Long Noncoding/genetics , Telomere Homeostasis/genetics , Telomere/genetics , Transcription, Genetic/genetics , Cell Line, Tumor , Chromosome Breakage , DNA Damage/genetics , DNA Replication/genetics , High-Throughput Nucleotide Sequencing , Humans , Neoplasms/genetics
9.
RNA ; 27(1): 106-121, 2021 01.
Article in English | MEDLINE | ID: mdl-33127860

ABSTRACT

Telomeric repeat-containing RNA (TERRA) molecules play important roles at telomeres, from heterochromatin regulation to telomerase activity control. In human cells, TERRA is transcribed from subtelomeric promoters located on most chromosome ends and associates with telomeres. The origin of mouse TERRA molecules is, however, unclear, as transcription from the pseudoautosomal PAR locus was recently suggested to account for the vast majority of TERRA in embryonic stem cells (ESC). Here, we confirm the production of TERRA from both the chromosome 18q telomere and the PAR locus in mouse embryonic fibroblasts, ESC, and various mouse cancer and immortalized cell lines, and we identify two novel sources of TERRA on mouse chromosome 2 and X. Using various approaches, we show that PAR-TERRA molecules account for the majority of TERRA transcripts, displaying an increase of two to four orders of magnitude compared to the telomeric 18q transcript. Finally, we present a SILAC-based pull-down screen revealing a large overlap between TERRA-interacting proteins in human and mouse cells, including PRC2 complex subunits, chromatin remodeling factors, DNA replication proteins, Aurora kinases, shelterin complex subunits, Bloom helicase, Coilin, and paraspeckle proteins. Hence, despite originating from distinct genomic regions, mouse and human TERRA are likely to play similar functions in cells.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , RNA, Messenger/genetics , Telomere/chemistry , Transcriptome , Animals , Aurora Kinases/genetics , Aurora Kinases/metabolism , Cell Line, Tumor , Chromosomes, Mammalian/chemistry , Chromosomes, Mammalian/metabolism , Computational Biology/methods , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Regulatory Networks , Granulocyte Precursor Cells/cytology , Granulocyte Precursor Cells/metabolism , HeLa Cells , Humans , Mice , Monocytes/cytology , Monocytes/metabolism , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neurons/cytology , Neurons/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA, Messenger/classification , RNA, Messenger/metabolism , RNA-Binding Proteins/classification , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , RecQ Helicases/genetics , RecQ Helicases/metabolism , Shelterin Complex , Telomere/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
10.
Front Mol Biosci ; 6: 84, 2019.
Article in English | MEDLINE | ID: mdl-31552268

ABSTRACT

The human FANCM ATPase/translocase is involved in various cellular pathways including DNA damage repair, replication fork remodeling and R-loop resolution. Recently, reports from three independent laboratories have disclosed a previously unappreciated role for FANCM in telomerase-negative human cancer cells that maintain their telomeres through the Alternative Lengthening of Telomeres (ALT) pathway. In ALT cells, FANCM limits telomeric replication stress and damage, and, in turn, ALT activity by suppressing accumulation of telomeric R-loops and by regulating the action of the BLM helicase. As a consequence, FANCM inactivation leads to exaggerated ALT activity and ultimately cell death. The studies reviewed here not only unveil a novel function for human FANCM, but also point to this enzyme as a promising target for anti-ALT cancer therapy.

11.
Genetics ; 213(2): 431-447, 2019 10.
Article in English | MEDLINE | ID: mdl-31405990

ABSTRACT

A subset of cancers rely on telomerase-independent mechanisms to maintain their chromosome ends. The predominant "alternative lengthening of telomeres" pathway appears dependent on homology-directed repair (HDR) to maintain telomeric DNA. However, the molecular changes needed for cells to productively engage in telomeric HDR are poorly understood. To gain new insights into this transition, we monitored the state of telomeres during serial culture of fission yeast (Schizosaccharomyces pombe) lacking the telomerase recruitment factor Ccq1. Rad52 is loaded onto critically short telomeres shortly after germination despite continued telomere erosion, suggesting that recruitment of recombination factors is not sufficient to maintain telomeres in the absence of telomerase function. Instead, survivor formation coincides with the derepression of telomeric repeat-containing RNA (TERRA). In this context, degradation of TERRA associated with the telomere in the form of R-loops drives a severe growth crisis, ultimately leading to a novel type of survivor with linear chromosomes and altered cytological telomere characteristics, including the loss of the shelterin component Rap1 (but not the TRF1/TRF2 ortholog, Taz1) from the telomere. We demonstrate that deletion of Rap1 is protective in this context, preventing the growth crisis that is otherwise triggered by degradation of telomeric R-loops in survivors with linear chromosomes. These findings suggest that upregulation of telomere-engaged TERRA, or altered recruitment of shelterin components, can support telomerase-independent telomere maintenance.


Subject(s)
Schizosaccharomyces pombe Proteins/genetics , Telomere Homeostasis/genetics , Telomere Shortening/genetics , Telomere-Binding Proteins/genetics , Telomere/genetics , DNA, Fungal/chemistry , DNA, Fungal/genetics , DNA-Binding Proteins/genetics , RNA, Fungal/chemistry , RNA, Fungal/genetics , Recombinational DNA Repair/genetics , Schizosaccharomyces/genetics , Shelterin Complex , Telomerase/genetics
12.
Nat Commun ; 10(1): 2253, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31138795

ABSTRACT

Telomerase negative immortal cancer cells elongate telomeres through the Alternative Lengthening of Telomeres (ALT) pathway. While sustained telomeric replicative stress is required to maintain ALT, it might also lead to cell death when excessive. Here, we show that the ATPase/translocase activity of FANCM keeps telomeric replicative stress in check specifically in ALT cells. When FANCM is depleted in ALT cells, telomeres become dysfunctional, and cells stop proliferating and die. FANCM depletion also increases ALT-associated marks and de novo synthesis of telomeric DNA. Depletion of the BLM helicase reduces the telomeric replication stress and cell proliferation defects induced by FANCM inactivation. Finally, FANCM unwinds telomeric R-loops in vitro and suppresses their accumulation in cells. Overexpression of RNaseH1 completely abolishes the replication stress remaining in cells codepleted for FANCM and BLM. Thus, FANCM allows controlled ALT activity and ALT cell proliferation by limiting the toxicity of uncontrolled BLM and telomeric R-loops.


Subject(s)
DNA Helicases/genetics , DNA Replication/genetics , RecQ Helicases/genetics , Telomere Homeostasis/genetics , Telomere/metabolism , Cell Death/genetics , Cell Line, Tumor , Cell Proliferation/genetics , DNA Helicases/metabolism , HEK293 Cells , HeLa Cells , Humans , RecQ Helicases/metabolism , Ribonuclease H/genetics , Ribonuclease H/metabolism
13.
Nat Struct Mol Biol ; 25(2): 147-153, 2018 02.
Article in English | MEDLINE | ID: mdl-29358759

ABSTRACT

The shelterin protein TRF2 assembles protective T loops at chromosome ends by stimulating intramolecular invasion of the telomeric G-rich single-stranded DNA (ssDNA) overhang into the duplex telomeric array. The other shelterin factor, TRF1, is thought to mainly facilitate telomeric dsDNA replication without directly participating in end protection. Here we show that in vitro human TRF2 stimulates invasion of G-rich TERRA-like RNA into telomeric dsDNA, leading to formation of telomeric RNA-DNA hybrids (telR loops). The N-terminal basic domain of TRF2 binds to TERRA-like RNA and enables TRF2 to promote efficient RNA invasion. TRF1, through its N-terminal acidic domain, counteracts TRF2-mediated RNA invasion but not ssDNA invasion. In vivo, when TRF1 is depleted or replaced with a variant lacking the acidic domain, TRF2 induces formation of telR loops, which in turn cause telomere loss. Hence, uncontrolled TRF2 threatens telomere integrity, and TRF1 directly supports end protection by suppressing harmful telR loops.


Subject(s)
Telomeric Repeat Binding Protein 1/chemistry , Telomeric Repeat Binding Protein 2/chemistry , Cell Line, Tumor , DNA/chemistry , DNA, Single-Stranded , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Oligonucleotides/chemistry , Protein Binding , Protein Domains , RNA/metabolism , Telomere/chemistry
14.
RNA ; 23(2): 134-141, 2017 02.
Article in English | MEDLINE | ID: mdl-28096443

ABSTRACT

The binding of sequence-specific RNA-interacting proteins, such as the bacteriophage MS2 or PP7 coat proteins, to their corresponding target sequences has been extremely useful and widely used to visualize single mRNAs in vivo. However, introduction of MS2 stem-loops into yeast mRNAs has recently been shown to lead to the accumulation of RNA fragments, suggesting that the loops impair mRNA decay. This result was questioned, because fragment occurrence was mainly assessed using ensemble methods, and their cellular localization and its implications had not been addressed on a single transcript level. Here, we demonstrate that the introduction of either MS2 stem-loops (MS2SL) or PP7 stem-loops (PP7SL) can affect the processing and subcellular localization of mRNA. We use single-molecule fluorescence in situ hybridization (smFISH) to determine the localization of three independent mRNAs tagged with the stem-loop labeling systems in glucose-rich and glucose starvation conditions. Transcripts containing MS2SL or PP7SL display aberrant localization in both the nucleus and the cytoplasm. These defects are most prominent in glucose starvation conditions, with nuclear mRNA processing being altered and stem-loop fragments abnormally enriching in processing bodies (PBs). The mislocalization of SL-containing RNAs is independent of the presence of the MS2 or PP7 coat protein (MCP or PCP).


Subject(s)
Cell Nucleus/metabolism , Cytoplasm/metabolism , Inverted Repeat Sequences , RNA Processing, Post-Transcriptional , RNA, Fungal/metabolism , RNA, Messenger/metabolism , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cell Nucleus/drug effects , Cell Nucleus/ultrastructure , Cytoplasm/drug effects , Cytoplasm/ultrastructure , Glucose/metabolism , Glucose/pharmacology , In Situ Hybridization, Fluorescence , Levivirus/chemistry , Plasmids/chemistry , Plasmids/metabolism , RNA Stability , RNA, Fungal/chemistry , RNA, Fungal/genetics , RNA, Messenger/chemistry , RNA, Messenger/genetics , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Single Molecule Imaging , Staining and Labeling/methods , Transfection
15.
J Cell Sci ; 130(4): 767-778, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28062851

ABSTRACT

Cactins constitute a family of eukaryotic proteins broadly conserved from yeast to human and required for fundamental processes such as cell proliferation, genome stability maintenance, organismal development and immune response. Cactin proteins have been found to associate with the spliceosome in several model organisms, nevertheless their molecular functions await elucidation. Here we show that depletion of human cactin leads to premature sister chromatid separation, genome instability and cell proliferation arrest. Moreover, cactin is essential for efficient splicing of thousands of pre-mRNAs, and incomplete splicing of the pre-mRNA of sororin (also known as CDCA5), a cohesin-associated factor, is largely responsible for the aberrant chromatid separation in cactin-depleted cells. Lastly, cactin physically and functionally interacts with the spliceosome-associated factors DHX8 and SRRM2. We propose that cellular complexes comprising cactin, DHX8 and SRRM2 sustain precise chromosome segregation, genome stability and cell proliferation by allowing faithful splicing of specific pre-mRNAs. Our data point to novel pathways of gene expression regulation dependent on cactin, and provide an explanation for the pleiotropic dysfunctions deriving from cactin inactivation in distant eukaryotes.


Subject(s)
Carrier Proteins/metabolism , Chromatids/metabolism , DEAD-box RNA Helicases/metabolism , RNA Precursors/genetics , RNA Splicing Factors/metabolism , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Nucleus Shape , Cell Proliferation , Genomic Instability , HEK293 Cells , HeLa Cells , Humans , Introns/genetics , Protein Binding , RNA Precursors/metabolism
16.
EMBO Rep ; 17(7): 999-1012, 2016 07.
Article in English | MEDLINE | ID: mdl-27154402

ABSTRACT

Telomerase-mediated telomere elongation provides cell populations with the ability to proliferate indefinitely. Telomerase is capable of recognizing and extending the shortest telomeres in cells; nevertheless, how this mechanism is executed remains unclear. Here, we show that, in the fission yeast Schizosaccharomyces pombe, shortened telomeres are highly transcribed into the evolutionarily conserved long noncoding RNA TERRA A fraction of TERRA produced upon telomere shortening is polyadenylated and largely devoid of telomeric repeats, and furthermore, telomerase physically interacts with this polyadenylated TERRA in vivo We also show that experimentally enhanced transcription of a manipulated telomere promotes its association with telomerase and concomitant elongation. Our data represent the first direct evidence that TERRA stimulates telomerase recruitment and activity at chromosome ends in an organism with human-like telomeres.


Subject(s)
DNA-Binding Proteins/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Telomerase/metabolism , Telomere Homeostasis , Telomere/genetics , Telomere/metabolism , DNA-Binding Proteins/genetics , Gene Expression Regulation, Fungal , Genome, Fungal , Poly A , Protein Binding , Telomere Shortening , Transcription, Genetic
17.
FEBS Lett ; 589(18): 2417-23, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26213367

ABSTRACT

Mpn1 is an exoribonuclease that modifies the spliceosomal small nuclear RNA (snRNA) U6 by trimming its oligouridine tail and introducing a cyclic phosphate group (>p). Mpn1 deficiency induces U6 3' end misprocessing, accelerated U6 decay and pre-mRNA splicing defects. Mutations in the human MPN1 gene are associated with the genodermatosis Clericuzio-type poikiloderma with neutropenia (PN). Here we present the deep sequencing of the >p-containing transcriptomes of mpn1Δ fission yeast and PN cells. While in yeast U6 seems to be the only substrate of Mpn1, human Mpn1 also processes U6atac snRNA. PN cells bear unstable U6atac species with aberrantly long and oligoadenylated 3' ends. Our data corroborate the link between Mpn1 and snRNA stability suggesting that PN could derive from pre-mRNA splicing aberrations.


Subject(s)
Phosphoric Diester Hydrolases/metabolism , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Small Nuclear/chemistry , RNA, Small Nuclear/metabolism , Base Sequence , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Molecular Sequence Data , Neutropenia/genetics , Neutropenia/pathology , Phosphoric Diester Hydrolases/deficiency , RNA Splicing , RNA, Small Nuclear/genetics , Sequence Analysis, RNA , Skin Abnormalities/genetics , Skin Abnormalities/pathology
18.
RNA Biol ; 12(9): 938-41, 2015.
Article in English | MEDLINE | ID: mdl-26158306

ABSTRACT

Alternative Lengthening of Telomeres (ALT) mechanisms allow telomerase-negative immortal cells to buffer replicative telomere shortening. ALT is naturally active in a number of human cancers and might be selected upon telomerase inactivation. ALT is thought to operate through homologous recombination (HR) occurring between telomeric repeats from independent chromosome ends. Indeed, suppression of a number of HR factors impairs ALT cell proliferation. Yet, how HR is initiated at ALT telomeres remains elusive. Mounting evidence suggests that the long noncoding telomeric RNA TERRA renders ALT telomeres recombinogenic by forming RNA:DNA hybrids with the telomeric C-rich strand. TERRA and telomeric hybrids act in concert with a number of other factors, including the RNA endoribonuclease RNaseH1 and the single stranded DNA binding protein RPA. The functional interaction network built upon these different players seems indispensable for ALT telomere maintenance, and digging into the molecular details of this previously unappreciated network might open the way to novel avenues for cancer treatments.


Subject(s)
RNA, Long Noncoding/genetics , Telomere Homeostasis , Homologous Recombination , Humans , Models, Biological , Protein Binding , RNA, Long Noncoding/metabolism , Telomere-Binding Proteins/metabolism , Transcription, Genetic
19.
Trends Cell Biol ; 25(1): 29-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25257515

ABSTRACT

Long noncoding telomeric repeat-containing RNAs - TERRAs - are transcribed in a regulated manner from telomeres throughout eukaryotes. TERRA molecules consist of chromosome end-specific subtelomeric sequences and telomeric repeats at their 3' ends. Recent work suggests that TERRA sustains several important functions at chromosome ends. TERRA can regulate telomere length through modulation of exonuclease 1 and telomerase, it may promote recruitment of chromatin modifiers to damaged telomeres and thereby enable DNA end-processing, and it may promote telomere protein composition changes during cell cycle progression. Furthermore, telomere transcription regulates chromosome-end mobility within the nucleus. We review how TERRA, by regulated expression and by providing a molecular scaffold for various protein enzymes, can support a large variety of vital functions.


Subject(s)
RNA, Long Noncoding/physiology , Telomere Homeostasis/physiology , Telomere/physiology , Animals , Cell Cycle/physiology , DNA Replication/physiology , Humans , Transcription, Genetic/physiology
20.
EMBO J ; 34(1): 115-29, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25398909

ABSTRACT

The telomeric transcriptome comprises multiple long non-coding RNAs generated by transcription of linear chromosome ends. In a screening performed in Schizosaccharomyces pombe, we identified factors modulating the cellular levels of the telomeric transcriptome. Among these factors, Cay1 is the fission yeast member of the conserved family of Cactins, uncharacterized proteins crucial for cell growth and survival. In cay1∆ mutants, the cellular levels of the telomeric factor Rap1 are drastically diminished due to defects in rap1+ pre-mRNA splicing and Rap1 protein stability. cay1∆ cells accumulate histone H3 acetylated at lysine 9 at telomeres, which become transcriptionally desilenced, are over-elongated by telomerase and cause chromosomal aberrations in the cold. Overexpressing Rap1 in cay1+ deleted cells significantly reverts all telomeric defects. Additionally, cay1∆ mutants accumulate unprocessed Tf2 retrotransposon RNA through Rap1-independent mechanisms. Thus, Cay1 plays crucial roles in cells by ultimately harmonizing expression of transcripts originating from seemingly unrelated genomic loci.


Subject(s)
Chromosomes, Fungal/metabolism , Nuclear Proteins/metabolism , Schizosaccharomyces/metabolism , Telomere/metabolism , Transcription, Genetic/physiology , Chromosome Aberrations , Chromosomes, Fungal/genetics , Gene Deletion , Nuclear Proteins/genetics , Protein Stability , RNA Splicing/physiology , RNA, Fungal/genetics , RNA, Fungal/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroelements/physiology , Schizosaccharomyces/genetics , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Shelterin Complex , Telomere/genetics , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...